1
|
Ding Y, Fan B, Zhu C, Chen Z. Shared and Related Molecular Targets and Actions of Salicylic Acid in Plants and Humans. Cells 2023; 12:219. [PMID: 36672154 PMCID: PMC9856608 DOI: 10.3390/cells12020219] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/29/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Salicylic acid (SA) is a phenolic compound produced by all plants that has an important role in diverse processes of plant growth and stress responses. SA is also the principal metabolite of aspirin and is responsible for many of the anti-inflammatory, cardioprotective and antitumor activities of aspirin. As a result, the number of identified SA targets in both plants and humans is large and continues to increase. These SA targets include catalases/peroxidases, metabolic enzymes, protein kinases and phosphatases, nucleosomal and ribosomal proteins and regulatory and signaling proteins, which mediate the diverse actions of SA in plants and humans. While some of these SA targets and actions are unique to plants or humans, many others are conserved or share striking similarities in the two types of organisms, which underlie a host of common biological processes that are regulated or impacted by SA. In this review, we compare shared and related SA targets and activities to highlight the common nature of actions by SA as a hormone in plants versus a therapeutic agent in humans. The cross examination of SA targets and activities can help identify new actions of SA and better explain their underlying mechanisms in plants and humans.
Collapse
Affiliation(s)
- Yuanyuan Ding
- College of Life Sciences, China Jiliang University, Hangzhou 310018, China
| | - Baofang Fan
- Department of Botany and Plant Pathology and Purdue Center for Plant Biology, Purdue University, West Lafayette, IN 47907-2054, USA
| | - Cheng Zhu
- College of Life Sciences, China Jiliang University, Hangzhou 310018, China
| | - Zhixiang Chen
- College of Life Sciences, China Jiliang University, Hangzhou 310018, China
- Department of Botany and Plant Pathology and Purdue Center for Plant Biology, Purdue University, West Lafayette, IN 47907-2054, USA
| |
Collapse
|
2
|
Lee D, Kim JW, Lee CY, Oh J, Hwang SH, Jo M, Kim SA, Choi W, Noh JK, Yi DK, Song M, Kim HG, Cho JY. Guettarda crispiflora Vahl Methanol Extract Ameliorates Acute Lung Injury and Gastritis by Suppressing Src Phosphorylation. PLANTS (BASEL, SWITZERLAND) 2022; 11:3560. [PMID: 36559672 PMCID: PMC9784507 DOI: 10.3390/plants11243560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/07/2022] [Accepted: 12/14/2022] [Indexed: 06/17/2023]
Abstract
Many species in the genus Guettarda are known to exert anti-inflammatory effects and are used as traditional medicinal plants to treat various inflammatory symptoms. However, no studies on the inflammatory activities of Guettarda crispiflora Vahl have been reported. The aim of the study was to investigate in vitro and in vivo the anti-inflammatory effects of a methanol extract of Guettarda crispiflora Vahl (Gc-ME). To determine the anti-inflammatory activity of Gc-ME, lipopolysaccharide (LPS)-, poly(I:C)-, or Pam3CSK4-treated RAW264.7 cells, HCl/EtOH- and LPS-treated mice were employed for in vitro and in vivo tests. LPS-induced nitric oxide production in RAW264.7 cells was determined by Griess assays and cytokine gene expression in LPS-activated RAW264.7 cells, confirmed by RT- and real-time PCR. Transcriptional activation was evaluated by luciferase reporter gene assay. Target protein validation was assessed by Western blot analysis and cellular thermal shift assays (CETSA) with LPS-treated RAW264.7 and gene-transfected HEK293 cells. Using both a HCl/EtOH-induced gastritis model and an LPS-induced lung injury model, inflammatory states were checked by scoring or evaluating gastric lesions, lung edema, and lung histology. Phytochemical fingerprinting of Gc-ME was observed by using liquid chromatography-mass spectrometry. Nitric oxide production induced by LPS and Pam3CSK4 in RAW264.7 cells was revealed to be reduced by Gc-ME. The LPS-induced upregulation of iNOS, COX-2, IL-6, and IL-1β was also suppressed by Gc-ME treatment. Gc-ME downregulated the promotor activities of AP-1 and NF-κB triggered by MyD88- and TRIF induction. Upstream signaling proteins for NF-κB activation, namely, p-p50, p-p65, p-IκBα, and p-Src were all downregulated by Ch-EE. Moreover, Src was revealed to be directly targeted by Gc-ME. This extract, orally treated strongly, attenuated the inflammatory symptoms in HCl/EtOH-treated stomachs and LPS-treated lungs. Therefore, these results strongly imply that Guettarda crispiflora can be developed as a promising anti-inflammatory remedy with Src-suppressive properties.
Collapse
Affiliation(s)
- Dahae Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ji Won Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Chae Young Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jieun Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - So Hyun Hwang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Minkyeong Jo
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seung A Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Wooram Choi
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | | | - Dong-Keun Yi
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Minkyung Song
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Han Gyung Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Research Institute of Biomolecule Control, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Research Institute of Biomolecule Control, Sungkyunkwan University, Suwon 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
3
|
Kim JW, Kwon KW, Kim MY, Cho JY. Potentilla paradoxa Nutt. Ethanol Extract Exhibits Anti-Inflammatory Effects by Suppression of the Src/NF-κB Signaling Pathway. PLANTS 2022; 11:plants11131750. [PMID: 35807703 PMCID: PMC9269291 DOI: 10.3390/plants11131750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 12/02/2022]
Abstract
Inflammation is an immune response that protects against harmful stimuli. However, severe inflammation can cause many diseases, such as diabetes, cancer, and arthritis. In this study, we examined the anti-inflammatory efficacy and mechanism of Potentilla paradoxa Nutt. ethanol extract (Pp-EE) as a new strategy for controlling the inflammatory response. Cellular activities and the molecular target of Pp-EE were identified in RAW264.7 cells and HEK293T cells. The effect of Pp-EE was analyzed using the Griess assay, the luciferase assay, reverse transcription-polymerase chain reaction, and Western blotting. To evaluate the in vivo effects, an HCl/EtOH-induced gastritis mouse model was used. NO production and pro-inflammatory gene (iNOS, COX-2, and TNF-α) mRNA levels were decreased by Pp-EE in a concentration-dependent manner without showing cytotoxicity. The activation of the transcription factor, particularly NF-κB, was effectively suppressed by Pp-EE. It was also found that Pp-EE directly inhibits the activation of Src in lipopolysaccharide (LPS)-treated RAW264.7 cells and in Src-overexpressed HEK293 cells by Western blotting analysis and cellular thermal shift assay. Experiments in the gastritis mouse model indicated that Pp-EE suppresses HCl/EtOH-induced gastric lesions, the expression levels of COX-2, IL-6, and TNF-α, and the phosphorylation of p65, p50, and Src. Taken together, these results suggest that Pp-EE can be applied as an anti-inflammatory remedy with a Src/NF-κB inhibitory property.
Collapse
Affiliation(s)
- Ji Won Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (J.W.K.); (K.W.K.)
| | - Ki Woong Kwon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (J.W.K.); (K.W.K.)
| | - Mi-Yeon Kim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Korea
- Correspondence: (M.-Y.K.); (J.Y.C.); Tel.: +82-2-820-0458 (M.-Y.K.); +82-31-290-7868 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (J.W.K.); (K.W.K.)
- Research Institute of Biomolecule Control and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Correspondence: (M.-Y.K.); (J.Y.C.); Tel.: +82-2-820-0458 (M.-Y.K.); +82-31-290-7868 (J.Y.C.)
| |
Collapse
|
4
|
Ohtsuki Y, Sanoh S, Santoh M, Ejiri Y, Ohta S, Kotake Y. Inhibition of cytochrome P450 3A protein degradation and subsequent increase in enzymatic activity through p38 MAPK activation by acetaminophen and salicylate derivatives. Biochem Biophys Res Commun 2018; 509:287-293. [PMID: 30587336 DOI: 10.1016/j.bbrc.2018.12.124] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 12/17/2018] [Indexed: 12/24/2022]
Abstract
Cytochrome P450 (CYP) 3A4 plays an important role in drug metabolism. Although transcriptional regulation of CYP3A expression by chemicals has been comprehensively studied, its post-translational regulation is not fully understood. We previously reported that acetaminophen (APAP) caused accumulation of functional CYP3A protein via inhibition of CYP3A protein degradation through reduction of glycoprotein 78 (gp78), an E3 ligase of the ubiquitin proteasome system. Furthermore, N-acetyl-m-aminophenol, a regioisomer of APAP causes CYP3A protein accumulation, whereas p-acetamidobezoic acid, in which a hydroxy group of APAP was substituted for a carboxy group, did not lead to the same effects. However, the mechanism underlying the reduction of gp78 protein expression by APAP has not yet been elucidated. In this study, we selected 32 compounds including a phenolic hydroxyl group such as APAP and explored the compounds that increased CYP3A enzyme activity to analyze their common mechanism. Four compounds, including salicylate, increased CYP3A enzyme activity and led to the accumulation of functional CYP3A protein similarly to APAP. APAP and salicylate activate p38 mitogen-activated protein kinase (p38 MAPK). gp78 is known to be phosphorylated by p38 MAPK; so, we investigated the relationship between p38 MAPK and CYP3A. APAP activated p38 MAPK, decreased gp78 protein expression, and subsequently induced CYP3A protein expression in a time-dependent manner. When SB203580, a p38 MAPK inhibitor, was co-administered with APAP, the inhibitory effects of APAP on CYP3A protein degradation were suppressed. In this study, we demonstrated the involvement of the p38 MAPK-gp78 pathway in suppressing CYP3A protein degradation by APAP. Salicylate derivatives may also suppress the CYP3A protein degradation.
Collapse
Affiliation(s)
- Yuya Ohtsuki
- Faculty of Pharmaceutical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Seigo Sanoh
- Faculty of Pharmaceutical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan; Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| | - Masataka Santoh
- Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Yoko Ejiri
- Molding Component Business Department, New Business Development Division, Kuraray Co., Ltd., 1-1-3 Otemachi, Chiyoda-ku, Tokyo, 100-8115, Japan
| | - Shigeru Ohta
- Faculty of Pharmaceutical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan; Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan; Wakayama Medical University, 811-1 Kimiidera, Wakayama, 641-8509, Japan
| | - Yaichiro Kotake
- Faculty of Pharmaceutical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan; Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| |
Collapse
|
5
|
Prescott JA, Cook SJ. Targeting IKKβ in Cancer: Challenges and Opportunities for the Therapeutic Utilisation of IKKβ Inhibitors. Cells 2018; 7:cells7090115. [PMID: 30142927 PMCID: PMC6162708 DOI: 10.3390/cells7090115] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 08/15/2018] [Accepted: 08/19/2018] [Indexed: 02/08/2023] Open
Abstract
Deregulated NF-κB signalling is implicated in the pathogenesis of numerous human inflammatory disorders and malignancies. Consequently, the NF-κB pathway has attracted attention as an attractive therapeutic target for drug discovery. As the primary, druggable mediator of canonical NF-κB signalling the IKKβ protein kinase has been the historical focus of drug development pipelines. Thousands of compounds with activity against IKKβ have been characterised, with many demonstrating promising efficacy in pre-clinical models of cancer and inflammatory disease. However, severe on-target toxicities and other safety concerns associated with systemic IKKβ inhibition have thus far prevented the clinical approval of any IKKβ inhibitors. This review will discuss the potential reasons for the lack of clinical success of IKKβ inhibitors to date, the challenges associated with their therapeutic use, realistic opportunities for their future utilisation, and the alternative strategies to inhibit NF-κB signalling that may overcome some of the limitations associated with IKKβ inhibition.
Collapse
Affiliation(s)
- Jack A Prescott
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| | - Simon J Cook
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK.
| |
Collapse
|
6
|
Wang W, Wu L, Li Q, Zhang Z, Xu L, Lin C, Gao L, Zhao K, Liang F, Zhang Q, Zhou M, Jiang W. Madecassoside prevents acute liver failure in LPS/D-GalN-induced mice by inhibiting p38/NF-κB and activating Nrf2/HO-1 signaling. Biomed Pharmacother 2018; 103:1137-1145. [PMID: 29715757 DOI: 10.1016/j.biopha.2018.04.162] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 04/23/2018] [Accepted: 04/23/2018] [Indexed: 12/30/2022] Open
Abstract
Madecassoside (MA), a triterpenoid saponin isolated from Centella asiatica, exerts various pharmacological activities including antioxidative and anti-inflammatory effects. The aim of this study was to explore the protective effect of MA in the treatment of lipopolysaccharide (LPS) and D-galactosamine (D-GalN)-induced acute liver failure(ALF) in mice. We hypothesized that MA administration may decrease the degree of liver injury caused by LPS/D-GalN. In this study, we investigated this hypothesis by treating a mouse model of LPS/D-GalN-induced liver injury with MA. Our study demonstrated that MA (20 mg/kg and 40 mg/kg) treatment for 10 days attenuated LPS/D-GalN-induced liver injury by protecting liver function, suppressing the production of inflammatory cytokines such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6, and recovering antioxidant enzyme activity. MA also significantly suppressed LPS-stimulated protein levels of inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX)-2 by blocking the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and eukaryotic transcription factor nuclear factor-kappa B (NF-κB). In addition, MA treatment enhanced protein levels of heme oxygenase (HO)-1 and anti-oxidant enzymes (superoxide dismutase, catalase, and glutathione peroxidase) through the upregulation of nuclear factor E2-related factor 2 (Nrf2) in LPS-stimulated liver injury. These results suggest that MA is a promising agent for the treatment of LPS/D-GalN-induced liver injury that could serve as a candidate for the development of a hepatoprotective drug against ALF.
Collapse
Affiliation(s)
- Wei Wang
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Lingling Wu
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Qing Li
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Zhuo Zhang
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Liba Xu
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Caixia Lin
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Ling Gao
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Kaili Zhao
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Fei Liang
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Qing Zhang
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Mei Zhou
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China
| | - Weizhe Jiang
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China.
| |
Collapse
|
7
|
Dihydrofisetin exerts its anti-inflammatory effects associated with suppressing ERK/p38 MAPK and Heme Oxygenase-1 activation in lipopolysaccharide-stimulated RAW 264.7 macrophages and carrageenan-induced mice paw edema. Int Immunopharmacol 2017; 54:366-374. [PMID: 29202300 DOI: 10.1016/j.intimp.2017.11.034] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/23/2017] [Accepted: 11/27/2017] [Indexed: 12/27/2022]
Abstract
Dihydrofisetin is a flavanonol derived from some edible wild herbs and traditional Chinese medicines. It has been found to possess many biological activities. However, the anti-inflammatory potential of Dihydrofisetin remains uncharacterized. The aim of the present study was to investigate the anti-inflammatory activity of Dihydrofisetin and its underlying mechanisms. We found that Dihydrofisetin dose-dependently inhibited lipopolysaccharide-induced productions of nitric oxide (NO) and prostaglandin E2 (PGE2) in RAW 264.7 macrophages, probably through suppressing the protein expressions of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). The expressions of pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6) and monocyte chemotactic protein (MCP-1) were also suppressed. We further demonstrated that Dihydrofisetin inhibited the activation of mitogen-activated protein kinases (MAPKs) pathway and phosphorylation of IκB-α whereas upregulated the expression of heme oxygenase-1 (HO-1). The in vivo carrageenan-induced mice paw edema study also indicated that treatment with 100 mg/kg of Dihydrofisetin could significantly inhibit carrageenan induced paw edema, decrease the levels of TNF-α, IL-6 and MDA, and increase the activity of GSH-Px in paw tissues. Taken together, Dihydrofisetin may act as a natural agent for treating inflammatory diseases by targeting MAPK, NF-κB and HO-1 pathways.
Collapse
|
8
|
Jevtić B, Djedović N, Stanisavljević S, Gašić U, Mišić D, Despotović J, Samardžić J, Miljković D, Timotijević G. Anti-encephalitogenic effects of cucumber leaf extract. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.07.060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
9
|
Madunić J, Horvat L, Majstorović I, Jodłowska I, Antica M, Matulić M. Sodium Salicylate Inhibits Urokinase Activity in MDA MB-231 Breast Cancer Cells. Clin Breast Cancer 2017; 17:629-637. [PMID: 28456486 DOI: 10.1016/j.clbc.2017.03.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/17/2017] [Accepted: 03/23/2017] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Sodium salicylate (NaS) is a derivate of acetylsalicylic acid or aspirin, used as a nonsteroidal anti-inflammatory drug for centuries, for its analgesic and anti-inflammatory effects. It was found to modulate different signaling pathways, in a cell-specific way. Here, we explore the effect of NaS on cell growth and urokinase activity in MDA MB-231 breast cancer cells. MATERIALS AND METHODS We analyzed the effect of NaS treatment on cell growth by flow cytometry and viability test. The transwell migration assay was used to study the migratory response of the cells. The gene expression was analyzed by qRT-PCR on RNA level and by Western blot analysis on protein level. Urokinase activity was assessed by caseinolysis. RESULTS Sublethal concentrations of NaS decreased cell growth and inhibited urokinase activity. The latter was a consequence of decrease in urokinase expression and increase in expression of its inhibitors. Analysis of signaling molecules revealed activation of transforming growth factor-β signaling, increase in master transcription factors for epithelial-mesenchymal transition and changes in integrin expression. CONCLUSIONS We propose that NaS causes partial cellular reprogramming through transforming growth factor-β signaling which, together with direct NaS influence, causes changes in expression in a set of genes involved in extracellular proteolysis. These data could be beneficial for the development of new therapeutic approaches in invasive breast cancer treatment.
Collapse
Affiliation(s)
- Josip Madunić
- Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Luka Horvat
- Faculty of Science, University of Zagreb, Zagreb, Croatia
| | | | - Iga Jodłowska
- Faculty of Science, University of Zagreb, Zagreb, Croatia
| | | | - Maja Matulić
- Faculty of Science, University of Zagreb, Zagreb, Croatia.
| |
Collapse
|
10
|
Zheng J, Son DJ, Lee HL, Lee HP, Kim TH, Joo JH, Ham YW, Kim WJ, Jung JK, Han SB, Hong JT. (E)-2-methoxy-4-(3-(4-methoxyphenyl)prop-1-en-1-yl)phenol suppresses ovarian cancer cell growth via inhibition of ERK and STAT3. Mol Carcinog 2017; 56:2003-2013. [PMID: 28277616 DOI: 10.1002/mc.22648] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 02/01/2017] [Accepted: 03/03/2017] [Indexed: 12/14/2022]
Abstract
In the present study, we synthesized several non-aldehyde analogues of (E)-2,4-bis(p-hydroxyphenyl)-2-butenal which showed anti-cancer effect. Interestingly, among the 16 compounds, we found that (E)-2-methoxy-4-(3-(4-methoxyphenyl)prop-1-en-1-yl)phenol (MMPP) showed the most significant anti-proliferative effect on PA-1 and SK-OV-3 ovarian epithelial cancer cells. MMPP treatment (0-15 µg/mL) induced apoptotic cell death, enhanced the expression of cleaved caspase-3, and cleaved caspase-9 in a concentration dependent manner. Notably, DNA binding activity of STAT3, phosphorylation of extracellular signal-regulated kinase (ERK) and p38 was significantly decreased by MMPP treatment. However, ERK siRNA augmented MMPP-induced inhibitory effect on cell growth rather than p38 siRNA or JNK siRNA. Moreover, combination treatment of MMPP with ERK inhibitor U0126 (10 µM) augmented MMPP-induced inhibitory effect on cell growth and DNA binding activity of STAT3, and enhanced expression of cleaved caspase-3 and cleaved caspase-9. In addition, STAT3 siRNA transfection augmented MMPP-induced cell growth inhibition. In PA-1 bearing xenograft mice model, MMPP (5 mg/kg) suppressed tumor growth significantly. Immunohistochemistry staining showed that the expression levels of p-ERK, PCNA, p-STAT3 were decreased while the expression level of caspase-3 was increased by MMPP treatment. Thus, MMPP may be a promising anti-cancer agent in ovarian epithelial cancer treatment.
Collapse
Affiliation(s)
- Jie Zheng
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Dong Ju Son
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Hye Lim Lee
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Hee Pom Lee
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Tae Hoon Kim
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jung Heun Joo
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Young Wan Ham
- Department of Chemistry, Utah Valley University 800 W, University Pkwy, Orem, Utah
| | - Wun Jae Kim
- College of Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jae Kyung Jung
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy & Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|
11
|
Lee JU, Chang HS, Lee HJ, Bae DJ, Son JH, Park JS, Choi JS, Hwang HG, Park CS. Association of interleukin-25 levels with development of aspirin induced respiratory diseases. Respir Med 2016; 123:71-78. [PMID: 28137499 DOI: 10.1016/j.rmed.2016.11.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 11/21/2016] [Accepted: 11/22/2016] [Indexed: 01/04/2023]
Abstract
BACKGROUND Aspirin-exacerbated respiratory diseases (AERD) are caused by ingestion of non-steroidal anti-inflammatory drugs and are characterized by acute bronchospasms and marked infiltration of eosinophils, the latter being attributable to altered synthesis of cysteinyl leukotrienes (LT) and prostaglandins (PG). Recently, the innate Th2 response is revealed to induce eosinophil infiltration in allergic inflammation, however the role of the innate Th2 response has not been studies in AERD. Thus, we evaluated the relationship between the innate Th2 cytokines including IL-25, thymic stromal lymphopoietin (TSLP) and IL-33 and the development of AERD. METHODS AND MATERIALS Plasma IL-25, IL-33, and TSLP levels were measured before and after aspirin challenge in subjects with AERD (n = 25) and aspirin-tolerant asthma (ATA, n = 25) by enzyme-linked immunosorbent assay (ELISA). Pre and post-aspirin challenge levels of LTC4 and PGD2 were measured using ELISA. RESULTS Basal plasma IL-25 levels were significantly higher in AERD group than in normal controls and in ATA group (p = 0.025 and 0.031, respectively). IL-33 and TSLP levels were comparable in the AERD and ATA groups. After the aspirin challenge, the IL-25 levels were markedly decreased in the ATA group (p = 0.024), while not changed in the AERD group. The post-challenge IL-25 levels of all asthmatic subjects were significantly correlated with aspirin challenge - induced declines in FEV1 (r = 0.357, p = 0.011), but not with basal and post challenge LTC4 and PGD2 levels. CONCLUSIONS IL-25 is associated with bronchospasm after aspirin challenge, possibly via mechanisms other than altered LTC4 and PGD2 production.
Collapse
Affiliation(s)
- Jong-Uk Lee
- Department of Interdisciplinary Program in Biomedical Science Major, Soonchunhyang Graduate School, Bucheon, Republic of Korea
| | - Hun Soo Chang
- Department of Interdisciplinary Program in Biomedical Science Major, Soonchunhyang Graduate School, Bucheon, Republic of Korea; Genome Research Center, Division of Allergy and Respiratory Medicine, Soonchunhyang University, Bucheon Hospital, Republic of Korea
| | - Hyeon Ju Lee
- Department of Interdisciplinary Program in Biomedical Science Major, Soonchunhyang Graduate School, Bucheon, Republic of Korea
| | - Da-Jeong Bae
- Department of Interdisciplinary Program in Biomedical Science Major, Soonchunhyang Graduate School, Bucheon, Republic of Korea
| | - Ji-Hye Son
- Department of Interdisciplinary Program in Biomedical Science Major, Soonchunhyang Graduate School, Bucheon, Republic of Korea
| | - Jong-Sook Park
- Genome Research Center, Division of Allergy and Respiratory Medicine, Soonchunhyang University, Bucheon Hospital, Republic of Korea
| | - Jae Sung Choi
- Division of Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University, Cheonan Hospital, Republic of Korea
| | - Hun Gyu Hwang
- Division of Allergy and Respiratory Medicine, Department of Internal Medicine, Soonchunhyang University, Gumi Hospital, Republic of Korea
| | - Choon-Sik Park
- Department of Interdisciplinary Program in Biomedical Science Major, Soonchunhyang Graduate School, Bucheon, Republic of Korea; Genome Research Center, Division of Allergy and Respiratory Medicine, Soonchunhyang University, Bucheon Hospital, Republic of Korea.
| |
Collapse
|
12
|
Li KK, Zhou X, Wong HL, Ng CF, Fu WM, Leung PC, Peng G, Ko CH. In vivo and in vitro anti-inflammatory effects of Zao-Jiao-Ci (the spine of Gleditsia sinensis Lam.) aqueous extract and its mechanisms of action. JOURNAL OF ETHNOPHARMACOLOGY 2016; 192:192-200. [PMID: 27401288 DOI: 10.1016/j.jep.2016.07.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 05/28/2016] [Accepted: 07/07/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zao-Jiao-Ci (ZJC), as the spine of Chinese Honey locust (Gleditsia sinensis Lam.), is traditionally used as Chinese medicine to reduce inflammation. AIM OF THE STUDY The present study aimed to investigate an anti-inflammatory effect of ZJC aqueous extract both in vitro and in vivo, as well as its underlying mechanisms. MATERIALS AND METHODS Anti-inflammatory effect of ZJC aqueous extract was evaluated by using carrageenan-induced paw edema in rats. In addition, the inhibitory effects of ZJC on nitric oxide production, intracellular reactive oxygen species production, pro-inflammatory mediator expression and prostaglandin E2 (PGE2) production were determined by using LPS-activated RAW 264.7 cells. The anti-oxidant activity of ZJC was assessed using 2,2'-azino-bis-3-ethylbenzthiazoline-6-sulfonic acid assay. RESULTS ZJC aqueous extract showed significant suppressive effect on paw edema in rats at 100mg/kg. Moreover, ZJC aqueous extract decreased the expression of cyclooxygenase (COX)-2 and significantly decreased the PGE2, tumor necrosis factor-α, interleukin (IL)-1β and IL-6 production in LPS-activated macrophages in dose-dependent manners. ZJC aqueous extract inhibited the mRNA expression of these inflammatory cytokines as well. Furthermore, ZJC aqueous extract was found as an anti-oxidant and could inhibit ROS production in the LPS-induced cells. CONCLUSIONS These findings show the potential of ZJC aqueous extract as a naturally occurring COX-2 inhibitor to reduce inflammation.
Collapse
Affiliation(s)
- Kai Kai Li
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China; Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| | - Xuelin Zhou
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China; Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| | - Hing Lok Wong
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China; Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| | - Chun Fai Ng
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China; Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| | - Wei Ming Fu
- Institute Guangzhou of Advanced Technology, Chinese Academy of Sciences, Guangzhou, China.
| | - Ping Chung Leung
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China; Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| | - Guiyuan Peng
- Department of ENT, Guangdong Province Traditional Chinese Medical Hospital, Guangzhou, China.
| | - Chun Hay Ko
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China; Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| |
Collapse
|
13
|
Jevtić B, Djedović N, Stanisavljević S, Despotović J, Miljković D, Timotijević G. Cucurbitacin E Potently Modulates the Activity of Encephalitogenic Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2016; 64:4900-7. [PMID: 27225664 DOI: 10.1021/acs.jafc.6b00951] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Cucurbitacin E (CucE) is a highly oxidized steroid consisting of a tetracyclic triterpene. It is a member of a Cucurbitacin family of biomolecules that are predominantly found in Cucurbitaceae plants. CucE has already been identified as a potent anti-inflammatory compound. Here, its effects on CD4(+) T helper (Th) cells and macrophages, as the major encephalitogenic cells in the autoimmunity of the central nervous system, were investigated. Production of major pathogenic Th cell cytokines: interferon-gamma and interleukin-17 were inhibited under the influence of CucE. The effects of CucE on CD4(+) T cells were mediated through the modulation of aryl hydrocarbon receptor, STAT3, NFκB, p38 MAPK, and miR-146 signaling. Further, production of nitric oxide and reactive oxygen species, as well as phagocytic ability, were inhibited in macrophages treated with CucE. These results imply that CucE possesses powerful antiencephalitogenic activity.
Collapse
Affiliation(s)
- Bojan Jevtić
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade , Belgrade, Serbia
| | - Neda Djedović
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade , Belgrade, Serbia
| | - Suzana Stanisavljević
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade , Belgrade, Serbia
| | - Jovana Despotović
- Laboratory for Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade , Belgrade, Serbia
| | - Djordje Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade , Belgrade, Serbia
| | - Gordana Timotijević
- Laboratory for Plant Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade , Belgrade, Serbia
| |
Collapse
|
14
|
Cameron AR, Logie L, Patel K, Bacon S, Forteath C, Harthill J, Roberts A, Sutherland C, Stewart D, Viollet B, Sakamoto K, McDougall G, Foretz M, Rena G. Investigation of salicylate hepatic responses in comparison with chemical analogues of the drug. Biochim Biophys Acta Mol Basis Dis 2016; 1862:1412-22. [PMID: 27130437 PMCID: PMC4894248 DOI: 10.1016/j.bbadis.2016.04.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 04/17/2016] [Accepted: 04/22/2016] [Indexed: 12/15/2022]
Abstract
Anti-hyperglycaemic effects of the hydroxybenzoic acid salicylate might stem from effects of the drug on mitochondrial uncoupling, activation of AMP-activated protein kinase, and inhibition of NF-κB signalling. Here, we have gauged the contribution of these effects to control of hepatocyte glucose production, comparing salicylate with inactive hydroxybenzoic acid analogues of the drug. In rat H4IIE hepatoma cells, salicylate was the only drug tested that activated AMPK. Salicylate also reduced mTOR signalling, but this property was observed widely among the analogues. In a sub-panel of analogues, salicylate alone reduced promoter activity of the key gluconeogenic enzyme glucose 6-phosphatase and suppressed basal glucose production in mouse primary hepatocytes. Both salicylate and 2,6 dihydroxybenzoic acid suppressed TNFα-induced IκB degradation, and in genetic knockout experiments, we found that the effect of salicylate on IκB degradation was AMPK-independent. Previous data also identified AMPK-independent regulation of glucose but we found that direct inhibition of neither NF-κB nor mTOR signalling suppressed glucose production, suggesting that other factors besides these cell signalling pathways may need to be considered to account for this response to salicylate. We found, for example, that H4IIE cells were exquisitely sensitive to uncoupling with modest doses of salicylate, which occurred on a similar time course to another anti-hyperglycaemic uncoupling agent 2,4-dinitrophenol, while there was no discernible effect at all of two salicylate analogues which are not anti-hyperglycaemic. This finding supports much earlier literature suggesting that salicylates exert anti-hyperglycaemic effects at least in part through uncoupling.
Collapse
Affiliation(s)
- Amy R Cameron
- Cardiovascular and Diabetes Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland DD1 9SY, United Kingdom
| | - Lisa Logie
- Cardiovascular and Diabetes Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland DD1 9SY, United Kingdom
| | - Kashyap Patel
- Cardiovascular and Diabetes Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland DD1 9SY, United Kingdom; MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, Scotland DD1 5EH, United Kingdom
| | - Sandra Bacon
- Cardiovascular and Diabetes Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland DD1 9SY, United Kingdom; James Hutton Institute, Invergowrie, Dundee, Scotland DD2 5DA, United Kingdom
| | - Calum Forteath
- Cardiovascular and Diabetes Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland DD1 9SY, United Kingdom
| | - Jean Harthill
- Cardiovascular and Diabetes Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland DD1 9SY, United Kingdom
| | - Adam Roberts
- Cardiovascular and Diabetes Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland DD1 9SY, United Kingdom
| | - Calum Sutherland
- Cardiovascular and Diabetes Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland DD1 9SY, United Kingdom
| | - Derek Stewart
- James Hutton Institute, Invergowrie, Dundee, Scotland DD2 5DA, United Kingdom; School of Life Sciences, Heriot-Watt University, Edinburgh, Scotland EH14 4AS, United Kingdom
| | - Benoit Viollet
- INSERM U1016, Institut Cochin, Paris 75014, France; CNRS UMR8104, Paris 75014, France; Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - Kei Sakamoto
- MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, Scotland DD1 5EH, United Kingdom; Nestlé Institute of Health Sciences SA, EPFL Innovation Park, Bâtiment G, 1015 Lausanne, Switzerland
| | - Gordon McDougall
- James Hutton Institute, Invergowrie, Dundee, Scotland DD2 5DA, United Kingdom
| | - Marc Foretz
- INSERM U1016, Institut Cochin, Paris 75014, France; CNRS UMR8104, Paris 75014, France; Université Paris Descartes, Sorbonne Paris Cité, Paris 75014, France
| | - Graham Rena
- Cardiovascular and Diabetes Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland DD1 9SY, United Kingdom.
| |
Collapse
|
15
|
Kong SK, Soo Kim B, Gi Uhm T, Soo Chang H, Sook Park J, Woo Park S, Park CS, Chung IY. Aspirin induces IL-4 production: augmented IL-4 production in aspirin-exacerbated respiratory disease. Exp Mol Med 2016; 48:e202. [PMID: 27534531 PMCID: PMC4686698 DOI: 10.1038/emm.2015.96] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 09/11/2015] [Indexed: 01/04/2023] Open
Abstract
Aspirin hypersensitivity is a hallmark of aspirin-exacerbated respiratory disease (AERD), a clinical syndrome characterized by the severe inflammation of the respiratory tract after ingestion of cyclooxygenase-1 inhibitors. We investigated the capacity of aspirin to induce interleukin-4 (IL-4) production in inflammatory cells relevant to AERD pathogenesis and examined the associated biochemical and molecular pathways. We also compared IL-4 production in peripheral blood mononuclear cells (PBMCs) from patients with AERD vs aspirin-tolerant asthma (ATA) upon exposure to aspirin. Aspirin induced IL-4 expression and activated the IL-4 promoter in a report assay. The capacity of aspirin to induce IL-4 expression correlated with its activity to activate mitogen-activated protein kinases, to form DNA-protein complexes on P elements in the IL-4 promoter and to synthesize nuclear factor of activated T cells, critical transcription factors for IL-4 transcription. Of clinical importance, aspirin upregulated IL-4 production twice as much in PBMCs from patients with AERD compared with PBMCs from patients with ATA. Our results suggest that IL-4 is an inflammatory component mediating intolerance reactions to aspirin, and thus is crucial for AERD pathogenesis.
Collapse
Affiliation(s)
- Su-Kang Kong
- Department of Bionano Technology, Hanyang University, Ansan, Republic of Korea
| | - Byung Soo Kim
- Department of Bionano Technology, Hanyang University, Ansan, Republic of Korea
| | - Tae Gi Uhm
- Department of Molecular and Life Sciences, College of Science and Technology, Hanyang University, Ansan, Republic of Korea
| | - Hun Soo Chang
- Soonchunhyang Medical Science Research Institute, College of Medicine, Soonchunhyang University, 22, Soonchunhyang-ro, Asan, Chungcheongnam-do, Republic of Korea
| | - Jong Sook Park
- Division of Allergy and Respiratory, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Gyeonggi-do, Republic of Korea
| | - Sung Woo Park
- Division of Allergy and Respiratory, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Gyeonggi-do, Republic of Korea
| | - Choon-Sik Park
- Division of Allergy and Respiratory, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Gyeonggi-do, Republic of Korea
| | - Il Yup Chung
- Department of Bionano Technology, Hanyang University, Ansan, Republic of Korea
- Department of Molecular and Life Sciences, College of Science and Technology, Hanyang University, Ansan, Republic of Korea
| |
Collapse
|
16
|
Sung B, Park S, Ha YM, Kim DH, Park CH, Jung KJ, Kim MS, Kim YJ, Kim MK, Moon JO, Yokozawa T, Kim ND, Yu BP, Chung HY. Salicylideneamino-2-thiophenol modulates nuclear factor-κB through redox regulation during the aging process. Geriatr Gerontol Int 2014; 15:211-9. [PMID: 25164597 DOI: 10.1111/ggi.12255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2013] [Indexed: 12/29/2022]
Abstract
AIM Many intracellular components have been implicated in the regulation of redox homeostasis, but homeostasis can be unbalanced by reactive species (RS), which also probably contribute to underlying inflammatory processes. Nuclear factor-κB (NF-κB) is a well-known redox-sensitive transcription factor that controls the genes responsible for regulating inflammation. METHODS In the present study, the authors investigated the effect of short-term salicylideneamino-2-thiophenol (SAL-2) administration on the modulation of pro-inflammatory NF-κB through redox regulation in aged rats. In addition, we compared the effects of SAL-2 and caloric restriction (CR) on inflammation and redox balance. The subjects were 24-month-old (old) Fischer 344 rats administered SAL-2 (10 mg/kg/day) by dietary supplementation or placed on a 30% restricted diet for 10 days, and 6-month-old (young) rats fed ad libitum for 10 days. RESULTS We found that NF-κB activation and the expressions of its related genes (vascular cell adhesion molecule-1, intercellular adhesion molecule-1, cyclooxygenase-2 and inducible nitric oxide synthase) were suppressed by SAL-2 supplementation in old CR rats to the levels observed in young rats. In addition, our molecular studies showed that the inhibitory effect of SAL-2 on the activation of NF-κB was mediated by the ability of SAL-2 to block the nuclear translocations of cytosolic thioredoxin and redox factor-1. CONCLUSION These findings strongly indicate that SAL-2 stabilizes age-related redox imbalance and modulates the signal transduction pathway involved in the age-associated molecular inflammatory process.
Collapse
Affiliation(s)
- Bokyung Sung
- Molecular Inflammation Research Center for Aging Intervention (MRCA), Pusan National University, Busan, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Jamal S, Cheriyan VT, Muthu M, Munie S, Levi E, Ashour AE, Pass HI, Wali A, Singh M, Rishi AK. CARP-1 functional mimetics are a novel class of small molecule inhibitors of malignant pleural mesothelioma cells. PLoS One 2014; 9:e89146. [PMID: 24598827 PMCID: PMC3943785 DOI: 10.1371/journal.pone.0089146] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 01/15/2014] [Indexed: 11/19/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an asbestos-related thoracic malignancy that is characterized by late metastases, and resistance to therapeutic modalities. The toxic side-effects of MPM therapies often limit their clinical effectiveness, thus necessitating development of new agents to effectively treat and manage this disease in clinic. CARP-1 functional mimetics (CFMs) are a novel class of compounds that inhibit growth of diverse cancer cell types. Here we investigated MPM cell growth suppression by the CFMs and the molecular mechanisms involved. CFM-1, -4, and -5 inhibited MPM cell growth, in vitro, in part by stimulating apoptosis. Apoptosis by CFM-4 involved activation of pro-apoptotic stress-activated protein kinases (SAPKs) p38 and JNK, elevated CARP-1 expression, cleavage of PARP1, and loss of the oncogene c-myc as well as mitotic cyclin B1. Treatments of MPM cells with CFM-4 resulted in depletion of NF-κB signaling inhibitor ABIN1 and Inhibitory κB (IκB)α and β, while increasing expression of pro-apoptotic death receptor (DR) 4 protein. CFM-4 enhanced expression of serine-phosphorylated podoplanin and cleavage of vimetin. CFMs also attenuated biological properties of the MPM cells by blocking their abilities to migrate, form colonies in suspension, and invade through the matrix-coated membranes. Both podoplanin and vimentin regulate processes of cell motility and invasion, and their expression often correlates with metastatic disease, and poor prognosis. The fact that phosphorylation of serines in the cytoplasmic domain of podoplanin interferes with processes of cellular motility, CFM-4-dependent elevated phosphorylated podoplanin and cleavage of vimentin underscore a metastasis inhibitory property of these compounds, and suggest that CFMs and/or their future analogs have potential as anti-MPM agents.
Collapse
Affiliation(s)
- Shazia Jamal
- John D. Dingell VA Medical Center, Wayne State University, Detroit, Michigan, United States of America
- Department of Oncology, Wayne State University, Detroit, Michigan, United States of America
| | - Vino T. Cheriyan
- John D. Dingell VA Medical Center, Wayne State University, Detroit, Michigan, United States of America
- Department of Oncology, Wayne State University, Detroit, Michigan, United States of America
| | - Magesh Muthu
- John D. Dingell VA Medical Center, Wayne State University, Detroit, Michigan, United States of America
- Department of Oncology, Wayne State University, Detroit, Michigan, United States of America
| | - Sara Munie
- John D. Dingell VA Medical Center, Wayne State University, Detroit, Michigan, United States of America
- Department of Oncology, Wayne State University, Detroit, Michigan, United States of America
| | - Edi Levi
- John D. Dingell VA Medical Center, Wayne State University, Detroit, Michigan, United States of America
- Department of Pathology, Wayne State University, Detroit, Michigan, United States of America
| | - Abdelkader E. Ashour
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Harvey I. Pass
- Division of Cardiothoracic Surgery, New York University Cancer Center, New York, United States of America
| | - Anil Wali
- John D. Dingell VA Medical Center, Wayne State University, Detroit, Michigan, United States of America
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, United States of America
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, United States of America
| | - Arun K. Rishi
- John D. Dingell VA Medical Center, Wayne State University, Detroit, Michigan, United States of America
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, United States of America
- Department of Oncology, Wayne State University, Detroit, Michigan, United States of America
- * E-mail:
| |
Collapse
|
18
|
Nohria A, Kinlay S, Buck JS, Redline W, Copeland-Halperin R, Kim S, Beckman JA. The effect of salsalate therapy on endothelial function in a broad range of subjects. J Am Heart Assoc 2014; 3:e000609. [PMID: 24390146 PMCID: PMC3959688 DOI: 10.1161/jaha.113.000609] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Inflammation is fundamental to the development of atherosclerosis. We examined the effect of anti‐inflammatory doses of salicylate on endothelium‐dependent vasodilation, a biomarker of cardiovascular risk, in a broad range of subjects. Methods and Results We performed a randomized, double‐blind, placebo‐controlled crossover trial evaluating the effects of 4 weeks of high‐dose salsalate (disalicylate) therapy on endothelium‐dependent flow‐mediated and endothelium‐independent vasodilation. Fifty‐eight subjects, including 17 with metabolic syndrome, 13 with atherosclerosis, and 28 healthy controls, were studied. Among all subjects, endothelium‐dependent flow‐mediated vasodilation decreased after salsalate compared with placebo therapy (P=0.01), whereas nitroglycerin‐mediated, endothelium‐independent vasodilation was unchanged (P=0.97). Endothelium‐dependent flow‐mediated vasodilation after salsalate therapy was impaired compared with placebo therapy in subjects with therapeutic salicylate levels (n=31, P<0.02) but not in subjects with subtherapeutic levels (P>0.2). Conclusions Salsalate therapy, particularly when therapeutic salicylate levels are achieved, impairs endothelium‐dependent vasodilation in a broad range of subjects. These data raise concern about the possible deleterious effects of anti‐inflammatory doses of salsalate on cardiovascular risk. Clinical Trial Registration URL: www.clinicaltrials.gov. Unique Identifiers: NCT00760019 and NCT00762827.
Collapse
Affiliation(s)
- Anju Nohria
- Cardiovascular Medicine, Department of Internal Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Aspirin-exacerbated respiratory disease is a clinical syndrome characterized by severe, persistent asthma, hyperplastic eosinophilic sinusitis with nasal polyps, and reactions to aspirin and other nonsteroidal antiinflammatory drugs that preferentially inhibit cyclooxygenase 1. The mechanisms behind the therapeutic effects of aspirin desensitization remain poorly understood. Recent studies suggest that the clinical benefits may occur through direct inhibition of tyrosine kinases and the signal transducer and activator of transcription 6 signaling pathway, which results in inhibition of interleukin 4 production. In this article, the current understanding of the mechanisms of aspirin desensitization is reviewed and future areas of investigation are discussed.
Collapse
Affiliation(s)
- Trever Burnett
- Department of Allergy and Immunology, National Jewish Medical and Research Center, University of Colorado, Denver, CO 80206, USA
| | | | | |
Collapse
|
20
|
Kollipara PS, Jeong HS, Han SB, Hong JT. (E)-2,4-bis(p-hydroxyphenyl)-2-butenal has an antiproliferative effect on NSCLC cells induced by p38 MAPK-mediated suppression of NF-κB and up-regulation of TNFRSF10B (DR5). Br J Pharmacol 2013; 168:1471-84. [PMID: 23082969 DOI: 10.1111/bph.12024] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Revised: 09/06/2012] [Accepted: 10/15/2012] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND AND PURPOSE The Maillard Reaction Products (MRPs) are known to be effective in chemoprevention. Here we focused on the anticancer effects of (E)-2,4-bis(p-hydroxyphenyl)-2-butenal (a MRP) on human non-small-cell lung cancer (NSCLC) cells and its mechanism of action. EXPERIMENTAL APPROACH We analysed the activity of (E)-2,4-bis(p-hydroxyphenyl)-2-butenal on NSCLC cells (NCI-H460 and A549) by use of Western blot analysis for major apoptotic proteins, MAPK, NF-κB and death receptor expression. We also used RT-PCR to determine its effects on death receptor mRNA expression, EMSA for effects on NF-κB DNA binding activity and colony formation assay for effects of inhibitors on (E)-2,4-bis(p-hydroxyphenyl)-2-butenal's actions. KEY RESULTS (E)-2,4-bis(p-hydroxyphenyl)-2-butenal induced a concentration (10-40 μg·mL⁻¹)- and time (30 min-72 h)-dependent inhibitory effect on the growth of NSCLC cells due to induction of apoptosis. Concomitantly, it significantly increased the expression of apoptotic proteins such as cleaved caspase-3, cleaved caspase-9, Bax and p53, but down-regulated the expression of anti-apoptotic proteins Bcl-2, cIAP1 and cIAP2. This effect was induced by up-regulation of MAPK and death receptor proteins TNFRSF12, TNFRSF10B and TNFRSF21, but suppression of NF-κB. Of the death receptors activated, only TNFRSF10B knock down with siRNA reversed the effect of (E)-2,4-bis(p-hydroxyphenyl)-2-butenal. Even though all the MAPKs were activated, only pretreatment with a p38 MAPK inhibitor reversed (E)-2,4-bis(p-hydroxyphenyl)-2-butenal-induced cell growth inhibition, increase in cleaved caspase-3, -9 and TNFRSF10B expression, and NF-κB inactivation. CONCLUSIONS AND IMPLICATIONS (E)-2,4-bis(p-hydroxyphenyl)-2-butenal induces apoptosis in NSCLC cells by p38 MAPK-mediated suppression of NF-κB and activation of TNFRSF10B, which then activates the caspase-3 and caspase-9 pathways.
Collapse
|
21
|
The proapoptotic effect of traditional and novel nonsteroidal anti-inflammatory drugs in mammalian and yeast cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:504230. [PMID: 23983899 PMCID: PMC3747411 DOI: 10.1155/2013/504230] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/08/2013] [Indexed: 12/16/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) have long been used to treat pain, fever, and inflammation. However, mounting evidence shows that NSAIDs, such as aspirin, have very promising antineoplastic properties. The chemopreventive, antiproliferative behaviour of NSAIDs has been associated with both their inactivation of cyclooxygenases (COX) and their ability to induce apoptosis via pathways that are largely COX-independent. In this review, the various proapoptotic pathways induced by traditional and novel NSAIDs such as phospho-NSAIDs, hydrogen sulfide-releasing NSAIDs and nitric oxide-releasing NSAIDs in mammalian cell lines are discussed, as well as the proapoptotic effects of NSAIDs on budding yeast which retains the hallmarks of mammalian apoptosis. The significance of these mechanisms in terms of the role of NSAIDs in effective cancer prevention is considered.
Collapse
|
22
|
Goldfine AB, Fonseca V, Jablonski KA, Chen YDI, Tipton L, Staten MA, Shoelson SE. Salicylate (salsalate) in patients with type 2 diabetes: a randomized trial. Ann Intern Med 2013; 159:1-12. [PMID: 23817699 PMCID: PMC4128629 DOI: 10.7326/0003-4819-159-1-201307020-00003] [Citation(s) in RCA: 182] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Short-duration studies show that salsalate improves glycemia in type 2 diabetes mellitus (T2DM). OBJECTIVE To assess 1-year efficacy and safety of salsalate in T2DM. DESIGN Placebo-controlled, parallel trial; computerized randomization and centralized allocation, with patients, providers, and researchers blinded to assignment. (ClinicalTrials.gov: NCT00799643). SETTING 3 private practices and 18 academic centers in the United States. PATIENTS Persons aged 18 to 75 years with fasting glucose levels of 12.5 mmol/L or less (≤225 mg/dL) and hemoglobin A1c (HbA1c) levels of 7.0% to 9.5% who were treated for diabetes. INTERVENTION 286 participants were randomly assigned (between January 2009 and July 2011) to 48 weeks of placebo (n = 140) or salsalate, 3.5 g/d (n = 146), in addition to current therapies, and 283 participants were analyzed (placebo, n = 137; salsalate, n = 146). MEASUREMENTS Change in hemoglobin A1c level (primary outcome) and safety and efficacy measures. RESULTS The mean HbA1c level over 48 weeks was 0.37% lower in the salsalate group than in the placebo group (95% CI, -0.53% to -0.21%; P < 0.001). Glycemia improved despite more reductions in concomitant diabetes medications in salsalate recipients than in placebo recipients. Lower circulating leukocyte, neutrophil, and lymphocyte counts show the anti-inflammatory effects of salsalate. Adiponectin and hematocrit levels increased more and fasting glucose, uric acid, and triglyceride levels decreased with salsalate, but weight and low-density lipoprotein cholesterol levels also increased. Urinary albumin levels increased but reversed on discontinuation; estimated glomerular filtration rates were unchanged. LIMITATION Trial duration and number of patients studied were insufficient to determine long-term risk-benefit of salsalate in T2DM. CONCLUSION Salsalate improves glycemia in patients with T2DM and decreases inflammatory mediators. Continued evaluation of mixed cardiorenal signals is warranted.
Collapse
Affiliation(s)
- Allison B Goldfine
- Joslin Diabetes Center and Harvard Medical School, Boston, Massachusetts 02215, USA.
| | | | | | | | | | | | | |
Collapse
|
23
|
Ashour AE, Jamal S, Cheryan VT, Muthu M, Zoheir KMA, Alafeefy AM, Abd-Allah AR, Levi E, Tarca AL, Polin LA, Rishi AK. CARP-1 functional mimetics: a novel class of small molecule inhibitors of medulloblastoma cell growth. PLoS One 2013; 8:e66733. [PMID: 23826121 PMCID: PMC3691183 DOI: 10.1371/journal.pone.0066733] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 05/09/2013] [Indexed: 01/31/2023] Open
Abstract
Medulloblastomas (MBs) constitute an aggressive class of intracranial pediatric tumors. Current multimodality treatments for MBs include surgery, ionizing radiation, and chemotherapy. Toxic side effects of therapies coupled with high incidence of recurrence and the metastatic spread warrant development of more effective, less toxic therapies for this disease. CARP-1/CCAR1 is a peri-nuclear phospho-protein that is a co-activator of the cell cycle regulatory anaphase promoting complex/cyclosome (APC/C) E3 ligase. CARP-1 functional mimetics (CFMs) are a novel class of small molecule compounds that interfere with CARP-1 binding with APC/C subunit APC-2, and suppress growth of a variety of cancer cells in part by promoting apoptosis. Here we investigated MB growth inhibitory potential of the CFMs and found that CFM-4 inhibits growth of MB cells in part by inducing CARP-1 expression, promoting PARP cleavage, activating pro-apoptotic stress-activated protein kinases (SAPK) p38 and JNK, and apoptosis. Gene-array-based analysis of the CFM-4-treated Daoy MB cells indicated down-regulation of a number of key cell growth and metastasis-promoting genes including cell motility regulating small GTP binding protein p21Rac1, and extracellular matrix metallopeptidase (MMP)-10. Moreover, CFM-4 treatment stimulated expression of a number of molecules such as neurotrophin (NTF)3, and NF-κB signaling inhibitors ABIN1 and 2 proteins. Overexpression of NTF3 resulted in reduced MB cell viability while knock-down of NTF3 interfered with CFM-4-dependent loss of viability. CFMs also attenuated biological properties of the MB cells by blocking their abilities to migrate, form colonies in suspension, and invade through the matrix-coated membranes. Together our data support anti-MB properties of CFM-4, and provide a proof-of-concept basis for further development of CFMs as potential anti-cancer agents for MBs.
Collapse
Affiliation(s)
- Abdelkader E. Ashour
- Department of Pharmacology and toxicology, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Shazia Jamal
- John D. Dingell Veterans Affairs Medical Center, Detroit, Michigan, United States of America
- Department of Oncology, Wayne State University, Detroit, Michigan, United States of America
| | - Vino T. Cheryan
- John D. Dingell Veterans Affairs Medical Center, Detroit, Michigan, United States of America
- Department of Oncology, Wayne State University, Detroit, Michigan, United States of America
| | - Magesh Muthu
- John D. Dingell Veterans Affairs Medical Center, Detroit, Michigan, United States of America
- Department of Oncology, Wayne State University, Detroit, Michigan, United States of America
| | - Khairy M. A. Zoheir
- Department of Pharmacology and toxicology, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
- Cell Biology Department, National Research Center, Dokki, Cairo, Egypt
| | - Ahmed M. Alafeefy
- Department of Pharmaceutical Chemistry, College of Pharmacy, Salman Bin Abdulaziz University, Alkharj, Kingdom of Saudi Arabia
| | - Adel R. Abd-Allah
- Department of Pharmacology and toxicology, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Edi Levi
- John D. Dingell Veterans Affairs Medical Center, Detroit, Michigan, United States of America
| | - Adi L. Tarca
- Department of Computer Science, Wayne State University, Detroit, Michigan, United States of America
| | - Lisa A. Polin
- Department of Oncology, Wayne State University, Detroit, Michigan, United States of America
| | - Arun K. Rishi
- John D. Dingell Veterans Affairs Medical Center, Detroit, Michigan, United States of America
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, United States of America
- Department of Oncology, Wayne State University, Detroit, Michigan, United States of America
- * E-mail:
| |
Collapse
|
24
|
Mahdi J, Al-Musayeib N, Mahdi E, Pepper C. Pharmacological Importance of Simple Phenolic Compounds on Inflammation, Cell Proliferation and Apoptosis with a Special Reference to β-D-Salicin and Hydroxybenzoic Acid. EUR J INFLAMM 2013. [DOI: 10.1177/1721727x1301100202] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Simple phenolic (SP) compounds are natural products that exhibit multiple pharmacological functions. The best known of these compounds is β-D-salicin, the first discovered phenolic glycoside and salicylic acid, or 2-hydroxybenzoic acid (2-HBA). Both of these compounds have attracted the interest of scientists in various interdisciplinary fields, including chemistry, pharmacology and medicine. Although β-D-salicin is found in various plants, it is often associated with willow, as it was first discovered in this species of plant. While the presence of glucose in β-D-salicin improves the physicochemical properties of the benzyl moiety, β-D-salicin itself does not have anti-inflammatory or anti-proliferative activity until it is metabolised into 2-HBA in the gastrointestinal tract and blood stream. Likewise, the majority of 2-acetoxybenzoic acid (2-ABA), or acetoxysalicylic acid also undergoes metabolic hydrolysis into 2-HBA. 2-HBA has been shown to play a role in modulating both inflammation and cancer partly through the inhibition of cyclooxygenase-2 (COX-2). It is now clear that 2-HBA most likely acts on the transcription factor NF-κB, which regulates the transcription of COX-2 thereby suppressing inflammation and cell proliferation and promoting apoptosis. Other phenolates, also exhibit anti-inflammation and anti-proliferation activities like the 4-hydroxybenzoate zinc (4-HBZn) complex, which was previously shown to preferentially inhibit COX-2 compared to 2-HBA and ASA. This review aims to collect all the available information related to β-D-salicin and other SP compounds in order to promote a new perspective of this interesting class of compounds and encourage further research into their pharmacological and clinical properties.
Collapse
Affiliation(s)
- J. Mahdi
- College of Medicine, Shaqra University, Riyadh, Saudi Arabia
| | - N. Al-Musayeib
- College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - E. Mahdi
- School of Medicine, Cardiff University, Cardiff, UK
| | - C. Pepper
- Institute of Cancer and Genetics, Cardiff University, Cardiff, UK
| |
Collapse
|
25
|
Intranasal delivery of nerve growth factor attenuates aquaporins-4-induced edema following traumatic brain injury in rats. Brain Res 2013. [DOI: 10.1016/j.brainres.2012.11.028] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
26
|
Tinsley HN, Grizzle WE, Abadi A, Keeton A, Zhu B, Xi Y, Piazza GA. New NSAID targets and derivatives for colorectal cancer chemoprevention. Recent Results Cancer Res 2013; 191:105-20. [PMID: 22893202 DOI: 10.1007/978-3-642-30331-9_6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Clinical and preclinical studies provide strong evidence that nonsteroidal anti-inflammatory drugs (NSAIDs) can prevent numerous types of cancers, especially colorectal cancer. Unfortunately, the depletion of physiologically important prostaglandins due to cyclooxygenase (COX) inhibition results in potentially fatal toxicities that preclude the long-term use of NSAIDs for cancer chemoprevention. While studies have shown an involvement of COX-2 in colorectal tumorigenesis, other studies suggest that a COX-independent target may be at least partially responsible for the antineoplastic activity of NSAIDs. For example, certain NSAID derivatives have been identified that do not inhibit COX-2 but have demonstrated efficacy to suppress carcinogenesis with potential for reduced toxicity. A number of alternative targets have also been reported to account for the tumor cell growth inhibitory activity of NSAIDs, including the inhibition of cyclic guanosine monophosphate phosphodiesterases (cGMP PDEs), generation of reactive oxygen species (ROS), the suppression of the apoptosis inhibitor protein, survivin, and others. Here, we review several promising mechanisms that are being targeted to develop safer and more efficacious NSAID derivatives for colon cancer chemoprevention.
Collapse
Affiliation(s)
- Heather N Tinsley
- Department of Biology, University of Montevallo, Montevallo, AL, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Adapala VJ, Ward M, Ajuwon KM. Adipose tissue biglycan as a potential anti-inflammatory target of sodium salicylate in mice fed a high fat diet. JOURNAL OF INFLAMMATION-LONDON 2012; 9:15. [PMID: 22533381 PMCID: PMC3536632 DOI: 10.1186/1476-9255-9-15] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 04/25/2012] [Indexed: 12/31/2022]
Abstract
Background Inflammation in adipose tissue (AT) during obesity causes impaired AT function. Although multiple extracellular matrix (ECM) proteins are expressed in AT their potential role in adipose tissue inflammation is unclear. Biglycan, a pro-inflammatory ECM gene, is highly enriched in adipose tissue. However, whether it is correlated with adipose tissue inflammation is unknown. We provide evidence in support of a strong association between biglycan expression and inflammatory status of adipose tissue. Methods C57BL6 mice were fed either a control (10% fat calories) or a high fat diet (HFD) (60% fat calories) for 8 weeks. Adipose tissue was analyzed for the expression of biglycan, IL-6 and TNFα. Biglycan knockout or wild type were also fed a high fat diet for 8 weeks and the expression of inflammatory genes in the mesenteric adipose tissue was examined. To test anti-inflammatory treatment on biglycan expression, a group of mice were fed either the low fat or high fat diet for eight weeks supplemented with either saline or sodium salicylate @ 25mg/100ml in their drinking water. Results Mice on HFD had an increase in ECM genes (BGN and COL1A1), inflammatory genes (IL-6 and TNFα) in both the subcutaneous and epididymal depots. However, correlation analysis only shows a positive correlation between biglycan, IL-6 and TNFα expression. In addition, lower expression of IL-6 and CD68 was found in the mesenteric adipose tissue of biglycan knockout mice compared to the wild type. Sodium salicylate treatment reduced subcutaneous adipose tissue expression of BGN, COL1A1, and COL6A1 and a concurrent downregulation of TNFα and IL-6 and TLR4 expression. Salicylate also lowered the serum TGFβ1 levels. Conclusion Biglycan expression correlates with adipose tissue inflammation, especially in the subcutaneous depot compared to the epididymal depot. This is supported by the greater effect of sodium salicylate in attenuating both inflammatory and ECM gene expression the subcutaneous adipose depot compared to the epididymal depot. These results show that inflammatory state may explain the induction of biglycan, and perhaps, other ECM genes in adipose tissue.
Collapse
Affiliation(s)
- Venkata J Adapala
- Department of Animal Sciences, Purdue University, West Lafayette, IN, 47907-2054, USA.
| | | | | |
Collapse
|
28
|
Pan Z, Wang Z, Yang H, Zhang F, Reinach PS. TRPV1 activation is required for hypertonicity-stimulated inflammatory cytokine release in human corneal epithelial cells. Invest Ophthalmol Vis Sci 2011; 52:485-93. [PMID: 20739465 DOI: 10.1167/iovs.10-5801] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE To determine whether hypertonic stress promotes increases in inflammatory cytokine release through transient receptor potential vanilloid channel type 1 (TRPV1) signaling pathway activation in human corneal epithelial cells (HCECs). METHODS Hyperosmotic medium was prepared by supplementing isotonic Ringers solution with sucrose. Ca2+ signaling was measured in fura2-AM-loaded HCECs using a single-cell fluorescence imaging system. Western blot analysis evaluated the phosphorylation status of EGFR, ERK, p38 MAPK, and nuclear factor (NF)-κB. ELISA assessed the effect of TRPV1 activation on the release of IL-6 and IL-8. RESULTS A 450 mOsm hypertonic stress elicited 2-fold Ca2+ transients that were suppressed by the TRPV1-selective antagonists capsazepine and JYL 1421. Such transients were enhanced by PGE2. Hypertonicity-induced EGF receptor (EGFR) transactivation was suppressed by preincubating HCECs with capsazepine, matrix metalloproteinase 1 (MMP1) inhibitor TIMP-1, broad-spectrum MMP inhibitor GM 6001, heparin-bound (HB)-EGF inhibitor CRM 197, or EGFR inhibitor AG 1478. ERK and p38 MAPK and NF-κB activation after EGFR transactivation occurred in tonicity and in a time-dependent manner. Hypertonicity-induced increases in IL-6 and IL-8 releases were suppressed by exposure to capsazepine, AG 1478, ERK inhibitor PD 98059, p38 inhibitor SB 203580, or NF-κB inhibitor PDTC. CONCLUSIONS Hypertonic stress-elicited TRPV1 channel stimulation mediates increases in a proinflammatory cytokine IL-6 and a chemoattractant IL-8 by eliciting EGFR transactivation, MAPK, and NF-κB activation. Selective drug modulation of either TRPV1 activity or its signaling mediators may yield a novel approach to suppressing inflammatory responses occurring in dry eye syndrome.
Collapse
Affiliation(s)
- Zan Pan
- Department of Biological Sciences, College of Optometry, State University of New York, New York, New York 10065, USA.
| | | | | | | | | |
Collapse
|
29
|
Wallace CS, Truskey GA. Direct-contact co-culture between smooth muscle and endothelial cells inhibits TNF-alpha-mediated endothelial cell activation. Am J Physiol Heart Circ Physiol 2010; 299:H338-46. [PMID: 20495148 PMCID: PMC2930383 DOI: 10.1152/ajpheart.01029.2009] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Accepted: 05/16/2010] [Indexed: 01/13/2023]
Abstract
We used a direct-contact endothelial cell-smooth muscle cell (EC-SMC) co-culture to examine whether quiescent SMCs regulate the EC inflammatory response to tumor necrosis factor (TNF)-alpha. ECs were cultured under static and physiological flow conditions. Compared with TNF-alpha-treated ECs in monoculture, TNF-alpha-treated ECs in co-culture had less NF-kappaB nuclear translocation; less intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and E-selectin surface protein expression; no change in TNF receptor expression, but greater Kruppel-like factor 2 (KLF2) gene expression. After flow preconditioning for 24 h at 15 dyne/cm(2), and exposure of ECs to flow and TNF-alpha for 4.5 h, ECs in co-culture had less ICAM-1, VCAM-1, and E-selectin surface protein expression. Exposure to flow greatly increased KLF2 gene expression levels in both EC cultures; as a result, ECs in co-culture and monoculture had similar levels of post-flow KLF2 gene expression. The reduced levels of TNF-alpha-induced adhesion molecule expression in co-culture required the presence of quiescent SMCs; adhesion to decellularized extracellular matrix (ECM) or co-culture with fibroblasts produced only a modest reduction in EC adhesion molecule expression. Furthermore, co-culture of quiescent SMCs and ECs on the opposite side of a 10-microm-thick porous membrane did not alter the TNF-alpha-mediated ICAM-1 surface protein expression. Although the ECM produced by SMCs plays some role in reducing TNF-alpha-mediated inflammation, these results suggest that the direct contact between ECs and quiescent SMCs is required to inhibit TNF-alpha-mediated activation.
Collapse
MESH Headings
- 3T3 Cells
- Animals
- Cell Adhesion
- Cell Communication
- Cell Proliferation
- Cellular Senescence
- Coculture Techniques
- E-Selectin/metabolism
- Endothelial Cells/immunology
- Endothelial Cells/metabolism
- Extracellular Matrix/metabolism
- Fibroblasts/metabolism
- Humans
- Inflammation Mediators/metabolism
- Intercellular Adhesion Molecule-1/metabolism
- Kruppel-Like Transcription Factors/metabolism
- Mice
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/immunology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/immunology
- Myocytes, Smooth Muscle/metabolism
- NF-kappa B/metabolism
- Pulsatile Flow
- Receptors, Tumor Necrosis Factor/metabolism
- Recombinant Proteins/metabolism
- Stress, Mechanical
- Time Factors
- Tumor Necrosis Factor-alpha/metabolism
- Vascular Cell Adhesion Molecule-1/metabolism
Collapse
Affiliation(s)
- Charles S Wallace
- Dept. of Biomedical Engineering, Duke Univ., Box 90281, Durham, NC 27708, USA
| | | |
Collapse
|
30
|
Fan JR, Huang TH, Wen CY, Shen TL, Li TK. Sodium salicylate acts through direct inhibition of phosphoinositide 3-kinase-like kinases to modulate topoisomerase-mediated DNA damage responses. Eur J Pharmacol 2010; 638:13-20. [DOI: 10.1016/j.ejphar.2010.04.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Revised: 04/04/2010] [Accepted: 04/12/2010] [Indexed: 12/31/2022]
|
31
|
Geller F, Schmidt C, Göttert M, Fronza M, Schattel V, Heinzmann B, Werz O, Flores EMM, Merfort I, Laufer S. Identification of rosmarinic acid as the major active constituent in Cordia americana. JOURNAL OF ETHNOPHARMACOLOGY 2010; 128:561-6. [PMID: 20149856 DOI: 10.1016/j.jep.2010.01.062] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Revised: 01/27/2010] [Accepted: 01/29/2010] [Indexed: 05/28/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Preparation from leaves of Cordia americana have been widely used in traditional medicine in South Brazil to treat wounds and various inflammations. AIM OF THE STUDY The objective of this work was to identify the effective compounds in the ethanolic extract prepared from the leaves of Cordia americana, which is used in traditional South Brazilian medicine as anti-inflammatory and wound healing remedy. MATERIALS AND METHODS Isolation and structure elucidation techniques were performed in order to identify the compounds of Cordia americana and HPLC analysis was used for the quantification. The major constituent and the ethanolic extract were investigated for inhibition of 5-lipoxygenase, p38alpha MAPK, TNFalpha release and NF-kappaB as well as in the fibroblast scratch assay. RESULTS Rosmarinic acid (1) was identified as the major compound with an amount of 8.44% in the ethanolic extract of the leaves of Cordia americana. The ethanolic extract as well as (1) exhibited the highest inhibitory effects on 5-lipoxygenase (IC(50)=0.69 and 0.97microg/mL, resp., IC50 of BWA4C as reference: 0.3microM) and p38alpha (IC50=3.25 and 1.16microg/mL, resp., IC50 of SB203580 as reference: 0.046microM) and moderate inhibitory effects on TNFalpha release. Slight effects were observed in the fibroblast scratch assay. CONCLUSIONS This study increases our knowledge on the effective compound in Cordia americana and supports its use in traditional medicine. We demonstrated for the first time pharmacological effects of Cordia americana and we provide evidences for a crucial role of rosmarinic acid as the major key player.
Collapse
Affiliation(s)
- F Geller
- Department of Pharmaceutical/Medicinal Chemistry, University of Tübingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Redondo S, Ruiz E, Gordillo-Moscoso A, Navarro-Dorado J, Ramajo M, Carnero M, Reguillo F, Rodriguez E, Tejerina T. Role of TGF-beta1 and MAP kinases in the antiproliferative effect of aspirin in human vascular smooth muscle cells. PLoS One 2010; 5:e9800. [PMID: 20339548 PMCID: PMC2842433 DOI: 10.1371/journal.pone.0009800] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Accepted: 12/26/2009] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND We aimed to test the antiproliferative effect of acetylsalicylic acid (ASA) on vascular smooth muscle cells (VSMC) from bypass surgery patients and the role of transforming growth factor beta 1 (TGF-beta1). METHODOLOGY/PRINCIPAL FINDINGS VSMC were isolated from remaining internal mammary artery from patients who underwent bypass surgery. Cell proliferation and DNA fragmentation were assessed by ELISA. Protein expression was assessed by Western blot. ASA inhibited BrdU incorporation at 2 mM. Anti-TGF-beta1 was able to reverse this effect. ASA (2 mM) induced TGF-beta1 secretion; however it was unable to induce Smad activation. ASA increased p38(MAPK) phosphorylation in a TGF-beta1-independent manner. Anti-CD105 (endoglin) was unable to reverse the antiproliferative effect of ASA. Pre-surgical serum levels of TGF-beta1 in patients who took at antiplatelet doses ASA were assessed by ELISA and remained unchanged. CONCLUSIONS/SIGNIFICANCE In vitro antiproliferative effects of aspirin (at antiinflammatory concentration) on human VSMC obtained from bypass patients are mediated by TGF-beta1 and p38(MAPK). Pre-surgical serum levels of TGF- beta1 from bypass patients who took aspirin at antiplatelet doses did not change.
Collapse
Affiliation(s)
- Santiago Redondo
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Emilio Ruiz
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid, Spain
| | | | - Jorge Navarro-Dorado
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Marta Ramajo
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid, Spain
| | - Manuel Carnero
- Servicio de Cirugía Cardiaca, Hospital Clinico Universitario San Carlos, Madrid, Spain
| | - Fernando Reguillo
- Servicio de Cirugía Cardiaca, Hospital Clinico Universitario San Carlos, Madrid, Spain
| | - Enrique Rodriguez
- Servicio de Cirugía Cardiaca, Hospital Clinico Universitario San Carlos, Madrid, Spain
| | - Teresa Tejerina
- Department of Pharmacology, School of Medicine, Universidad Complutense, Madrid, Spain
- * E-mail:
| |
Collapse
|
33
|
|
34
|
Li RWS, Seto SW, Au ALS, Kwan YW, Chan SW, Lee SMY, Tse CM, Leung GPH. Inhibitory effect of nonsteroidal anti-inflammatory drugs on adenosine transport in vascular smooth muscle cells. Eur J Pharmacol 2009; 612:15-20. [DOI: 10.1016/j.ejphar.2009.04.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2008] [Revised: 04/02/2009] [Accepted: 04/09/2009] [Indexed: 01/04/2023]
|
35
|
Schmidt C, Fronza M, Goettert M, Geller F, Luik S, Flores EMM, Bittencourt CF, Zanetti GD, Heinzmann BM, Laufer S, Merfort I. Biological studies on Brazilian plants used in wound healing. JOURNAL OF ETHNOPHARMACOLOGY 2009; 122:523-532. [PMID: 19429323 DOI: 10.1016/j.jep.2009.01.022] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2008] [Revised: 12/16/2008] [Accepted: 01/15/2009] [Indexed: 05/27/2023]
Abstract
AIM OF THE STUDY n-Hexanic and ethanolic extracts from twelve plants (Brugmansia suaveolens Brecht. et Presl., Eupatorium laevigatum Lam., Galinsoga parviflora Cav., Iresine herbstii Hook., Kalanchöe tubiflora Hamet-Ahti, Petiveria alliacea L., Pluchea sagittalis (Lam.) Cabrera, Piper regnellii DC., Schinus molle L., Sedum dendroideum Moç et Sessé ex DC., Waltheria douradinha St. Hill., Xanthium cavanillesii Schouw.) used in traditional South Brazilian medicine as wound healing agents were investigated in various biological assays, targeting different aspects in this complex process. MATERIALS AND METHODS The extracts were investigated on NF-kappaB DNA binding, p38alpha MAPK, TNF-alpha release, direct elastase inhibition and its release as well as on caspase-3. Fibroblasts migration to and proliferation into the wounded monolayers were evaluated in the scratch assay, the agar diffusion test for antibacterial and the MTT assay for cytotoxic effects. RESULTS The hydrophilic extracts from Galinsoga parviflora, Petiveria alliacea, Schinus molle, Waltheria douradinha and Xanthium cavanillesii as well as the lipophilic extract of Waltheria douradinha turned out to be the most active ones. CONCLUSIONS These results increase our knowledge on the wound healing effects of the investigated medicinal plants. Further studies are necessary to find out the effective secondary metabolites responsible for the observed effects.
Collapse
Affiliation(s)
- C Schmidt
- Department of Pharmaceutical Biology and Biotechnology, Institute for Pharmaceutical Sciences, University of Freiburg, Freiburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Thun MJ, Blackard B. Pharmacologic effects of NSAIDs and implications for the risks and benefits of long-term prophylactic use of aspirin to prevent cancer. Recent Results Cancer Res 2009; 181:215-21. [PMID: 19213571 DOI: 10.1007/978-3-540-69297-3_20] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
This paper briefly reviews the pharmacologic effects of nonsteroidal antiinflammatory drugs (NSAIDs) that influence the risks and benefits of using these drugs prophylactically for cancer. It describes the metabolism of arachidonic acid through the cyclooxygenase (COX) pathway, the physiologic functions ofprostanoids (prostaglandins, prostacyclin, and thromboxane A2) produced by this pathway, and the pharmacologic consequences of blocking the enzymatic activity of the two COX isoforms. We mention other proposed mechanisms by which NSAIDs may directly or indirectly affect non-COX pathways. The diverse pharmacologic effects of NSAIDs, when combined with the relatively low probability that an individual with average risk will develop any single type of cancer over a lifetime, severely limit the tolerance for toxicity if aspirin or related drugs are to be administered prophylactically to large numbers of otherwise healthy people. Further research is needed to identify a drug, dose, treatment regimen, and patient population(s) where the benefits of prophy- lactic treatment will exceed the risks. A singular advantage of aspirin over all other NSAIDs is the potential to combine reduced risk of certain cancers with cardiovascular benefit. However, many elements that are needed to achieve this remain unresolved.
Collapse
Affiliation(s)
- Michael J Thun
- Epidemiology and Surveillance Research, American Cancer Society, NW, Atlanta, GA 30303-1002, USA.
| | | |
Collapse
|
37
|
Fang X, Feng Y, Shi Z, Dai J. Alterations of cytokines and MAPK signaling pathways are related to the immunotoxic effect of perfluorononanoic acid. Toxicol Sci 2009; 108:367-76. [PMID: 19196829 DOI: 10.1093/toxsci/kfp019] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Perfluorononanoate (PFNA), a perfluorinated alkyl acid containing nine carbon chains, has been detected in abiotic and biotic matrices worldwide. Although a few studies have reported toxic effects of PFNA, little information of the mechanism has been offered. In this study, the effects of PFNA exposure on thymus and the related mechanisms were investigated. Male rats were orally dosed with 0, 1, 3, or 5 mg PFNA/kg/day for 14 days. A significant decrease of body weight and thymus weight were observed in the rats receiving 3 or 5 mg PFNA/kg/day. Histopathological examination revealed dose-dependent increases in thymocyte apoptosis. Rats receiving 3 or 5 mg PFNA/kg/day exhibited increased interleukin (IL)-1 and decreased IL-2 concentrations in sera, whereas elevated IL-4 and cortisol levels only occurred in the highest dose group. Quantitative real-time PCR indicated that expression of peroxisome proliferator-activated receptor alpha (PPAR-alpha) was increased in the thymi of all dosed rats, and a similar trend occurred for PPAR-gamma in the two highest dose groups. The mRNA levels of c-Jun NH(2)-terminal kinase (JNK), nuclear factor-kappa B, p65 subunit, and inhibitory protein IkappaBalpha were unchanged; however, increased and decreased mRNA levels of p38 kinase were found in rats exposed to 3 or 5 mg PFNA/kg/day, respectively. Decreased Bcl-2 mRNA levels were observed in rats receiving 5 mg PFNA/kg/day. A significant increase in protein levels of phospho-JNK was found in all PFNA-treated rats. Phospho-p38 was significantly enhanced in 1 and 3 mg PFNA/kg/day groups, whereas phospho-IkappaBalpha remained consistent in all rats studied. Together, these data suggested that apart from the activation of PPARs, PFNA exposure in rats lead to the alteration of serum cytokines, which subsequently activated mitogen-activated protein kinase signaling pathways and potentially modulated the immune system. Additionally, increased serum cortisol and decreased expression of Bcl-2 in thymus likely contributed to the PFNA-induced thymocyte apoptosis.
Collapse
Affiliation(s)
- Xuemei Fang
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | | | | | | |
Collapse
|
38
|
Tang W, Wang W, Zhang Y, Liu S, Liu Y, Zheng D. Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced chemokine release in both TRAIL-resistant and TRAIL-sensitive cells via nuclear factor kappa B. FEBS J 2008; 276:581-93. [PMID: 19120450 DOI: 10.1111/j.1742-4658.2008.06809.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis in a variety of tumour cells, but not in most normal cells, and has attracted considerable attention for its potential use in cancer therapy. Recently, increasing evidence has shown that TRAIL is involved in inflammation, although much of this evidence is controversial. In this article, it is shown that TRAIL induces CXCL2, CCL4 and CCL20 secretion in a nuclear factor kappa B-dependent manner. The dominant negative constructs of tumour necrosis factor receptor-associated death domain protein (TRADD) and tumour necrosis factor receptor-associated factor 2 are unable to block TRAIL-induced chemokine up-regulation, and the dominant negative construct of TRADD may even enhance TRAIL-triggered signals. Using small interfering RNA, receptor interacting protein has been demonstrated to be essential for TRAIL-induced chemokine release. Furthermore, it has been demonstrated that p38 mitogen-activated protein kinase is involved in TRAIL-induced chemokine release without any effects on nuclear factor kappa B activation, suggesting that some unknown transcription factors may be activated by TRAIL. Using a xenograft tumour model, it has been illustrated that TRAIL can also induce chemokine release in vivo. Although these chemokines induced by TRAIL are inflammatory chemokines, their functions are not restricted to inflammation and require further examination. Our results indicate that attention should be paid to the side-effects of TRAIL treatment, not only in TRAIL-resistant but also in TRAIL-sensitive tumour cells.
Collapse
Affiliation(s)
- Wanhu Tang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | | | | | | | | | | |
Collapse
|
39
|
Sapienza K, Bannister W, Balzan R. Mitochondrial involvement in aspirin-induced apoptosis in yeast. MICROBIOLOGY-SGM 2008; 154:2740-2747. [PMID: 18757807 DOI: 10.1099/mic.0.2008/017228-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
We have previously reported that aspirin induces apoptosis in manganese superoxide dismutase (MnSOD)-deficient Saccharomyces cerevisiae cells when cultivated on the non-fermentable carbon source ethanol. Here, we investigated the role of mitochondria in aspirin-induced apoptosis. We report that aspirin had an inhibitory effect on cellular respiration, and caused the release of most of the mitochondrial cytochrome c and a dramatic drop in the mitochondrial membrane potential (DeltaPsi(m)). Also, aspirin reduced the intracellular cytosolic pH in the MnSOD-deficient cells growing in ethanol medium, but this did not seem to be the initial trigger that committed these cells to aspirin-induced apoptosis. Furthermore, loss of DeltaPsi(m) was not required for aspirin-induced release of cytochrome c, since the initial release of cytochrome c occurred prior to the disruption of the DeltaPsi(m). It is thus possible that cytochrome c release does not involve the early onset of the mitochondrial permeability transition, but only an alteration of the permeability of the outer mitochondrial membrane.
Collapse
Affiliation(s)
- Karen Sapienza
- Department of Physiology and Biochemistry, University of Malta, Msida MSD 2080, Malta
| | - William Bannister
- Department of Physiology and Biochemistry, University of Malta, Msida MSD 2080, Malta
| | - Rena Balzan
- Department of Physiology and Biochemistry, University of Malta, Msida MSD 2080, Malta
| |
Collapse
|
40
|
An HJ, Rim HK, Lee JH, Suh SE, Lee JH, Kim NH, Choi IY, Jeong HJ, Kim IK, Lee JY, An NH, Kim HR, Um JY, Kim HM, Hong SH. Leonurus sibiricusinduces nitric oxide and tumor necrosis factor-α in mouse peritoneal macrophages. Can J Physiol Pharmacol 2008; 86:682-90. [DOI: 10.1139/y08-074] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Using mouse peritoneal macrophages, we have examined the mechanism by which Leonurus sibiricus (LS) regulates nitric oxide (NO) production. When LS was used in combination with recombinant interferon-γ (rIFN-γ), there was a marked cooperative induction of NO production; however, LS by itself had no effect on NO production. The increased production of NO from rIFN-γ plus LS-stimulated cells was almost completely inhibited by pretreatment with pyrrolidine dithiocarbamate (PDTC), an inhibitor of nuclear factor κB. Furthermore, treatment of peritoneal macrophages with rIFN-γ plus LS caused a significant increase in tumor necrosis factor-α (TNF-α) production. PDTC also decreased the effect of LS on TNF-α production significantly. Because NO and TNF-α play an important role in immune function and host defense, LS treatment could modulate several aspects of host defense mechanisms as a result of stimulation of the inducible nitric oxide synthase.
Collapse
Affiliation(s)
- Hyo-Jin An
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
- College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biochip Research Center, Hoseo University, Chungnam, Republic of Korea
- Division of Bio-Nano Chemistry, Wonkwang University, Iksan, Republic of Korea
- Department of Herb Science, Shinsung College, Chungnam, Republic of Korea
| | - Hong-Kun Rim
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
- College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biochip Research Center, Hoseo University, Chungnam, Republic of Korea
- Division of Bio-Nano Chemistry, Wonkwang University, Iksan, Republic of Korea
- Department of Herb Science, Shinsung College, Chungnam, Republic of Korea
| | - Jong-Hyun Lee
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
- College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biochip Research Center, Hoseo University, Chungnam, Republic of Korea
- Division of Bio-Nano Chemistry, Wonkwang University, Iksan, Republic of Korea
- Department of Herb Science, Shinsung College, Chungnam, Republic of Korea
| | - Se-Eun Suh
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
- College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biochip Research Center, Hoseo University, Chungnam, Republic of Korea
- Division of Bio-Nano Chemistry, Wonkwang University, Iksan, Republic of Korea
- Department of Herb Science, Shinsung College, Chungnam, Republic of Korea
| | - Ji-Hyun Lee
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
- College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biochip Research Center, Hoseo University, Chungnam, Republic of Korea
- Division of Bio-Nano Chemistry, Wonkwang University, Iksan, Republic of Korea
- Department of Herb Science, Shinsung College, Chungnam, Republic of Korea
| | - Na-Hyung Kim
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
- College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biochip Research Center, Hoseo University, Chungnam, Republic of Korea
- Division of Bio-Nano Chemistry, Wonkwang University, Iksan, Republic of Korea
- Department of Herb Science, Shinsung College, Chungnam, Republic of Korea
| | - In-Young Choi
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
- College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biochip Research Center, Hoseo University, Chungnam, Republic of Korea
- Division of Bio-Nano Chemistry, Wonkwang University, Iksan, Republic of Korea
- Department of Herb Science, Shinsung College, Chungnam, Republic of Korea
| | - Hyun-Ja Jeong
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
- College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biochip Research Center, Hoseo University, Chungnam, Republic of Korea
- Division of Bio-Nano Chemistry, Wonkwang University, Iksan, Republic of Korea
- Department of Herb Science, Shinsung College, Chungnam, Republic of Korea
| | - Il Kwang Kim
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
- College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biochip Research Center, Hoseo University, Chungnam, Republic of Korea
- Division of Bio-Nano Chemistry, Wonkwang University, Iksan, Republic of Korea
- Department of Herb Science, Shinsung College, Chungnam, Republic of Korea
| | - Ju-Young Lee
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
- College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biochip Research Center, Hoseo University, Chungnam, Republic of Korea
- Division of Bio-Nano Chemistry, Wonkwang University, Iksan, Republic of Korea
- Department of Herb Science, Shinsung College, Chungnam, Republic of Korea
| | - Nyeon-Hyoung An
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
- College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biochip Research Center, Hoseo University, Chungnam, Republic of Korea
- Division of Bio-Nano Chemistry, Wonkwang University, Iksan, Republic of Korea
- Department of Herb Science, Shinsung College, Chungnam, Republic of Korea
| | - Hyung-Ryong Kim
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
- College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biochip Research Center, Hoseo University, Chungnam, Republic of Korea
- Division of Bio-Nano Chemistry, Wonkwang University, Iksan, Republic of Korea
- Department of Herb Science, Shinsung College, Chungnam, Republic of Korea
| | - Jae-Young Um
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
- College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biochip Research Center, Hoseo University, Chungnam, Republic of Korea
- Division of Bio-Nano Chemistry, Wonkwang University, Iksan, Republic of Korea
- Department of Herb Science, Shinsung College, Chungnam, Republic of Korea
| | - Hyung-Min Kim
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
- College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biochip Research Center, Hoseo University, Chungnam, Republic of Korea
- Division of Bio-Nano Chemistry, Wonkwang University, Iksan, Republic of Korea
- Department of Herb Science, Shinsung College, Chungnam, Republic of Korea
| | - Seung-Heon Hong
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang University, Iksan, Jeonbuk 570-749, Republic of Korea
- College of Oriental Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biochip Research Center, Hoseo University, Chungnam, Republic of Korea
- Division of Bio-Nano Chemistry, Wonkwang University, Iksan, Republic of Korea
- Department of Herb Science, Shinsung College, Chungnam, Republic of Korea
| |
Collapse
|
41
|
Kim JM, Kim JS, Yoo H, Choung MG, Sung MK. Effects of black soybean [Glycine max (L.) Merr.] seed coats and its anthocyanidins on colonic inflammation and cell proliferation in vitro and in vivo. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2008; 56:8427-33. [PMID: 18710248 DOI: 10.1021/jf801342p] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Anti-inflammatory and antiproliferative activities of anthocyanidins and anthocyanin-rich black soybean seed coats were studied in HT-29 human colon adenocarcinoma cells and carcinogen-treated F344 rats, respectively. Cyanidin and delphinidin significantly inhibited cell growth at concentrations of >or=1 microM. Anthocyanidins suppressed cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) mRNAs in TPA-stimulated HT-29 cells. Both yellow and black soybean seed coat supplementation (10%, w/w) did not significantly reduce the number of aberrant crypt foci (ACF), although a modest decrease in the number of ACF was observed in animals fed soybean seed coats. The colonic COX-2 mRNA level was suppressed in rats fed both soybean seed coat diet. The plasma prostaglandin E 2 (PGE 2) level was reduced only in rats fed black soybean seed coats. No difference was observed in either colonic iNOS mRNA or plasma nitric oxide level. These results indicate that anthocyanidins are possible anti-inflammatory agents; however, further studies are required to determine required intake levels in vivo to exert antitumor effect.
Collapse
Affiliation(s)
- Jung-Mi Kim
- Department of Food and Nutrition, Sookmyung Women's University, Seoul 140-742, Korea
| | | | | | | | | |
Collapse
|
42
|
Wang J, Wang G, Ansari GAS, Khan MF. Activation of oxidative stress-responsive signaling pathways in early splenotoxic response of aniline. Toxicol Appl Pharmacol 2008; 230:227-34. [PMID: 18420242 PMCID: PMC2614137 DOI: 10.1016/j.taap.2008.02.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Revised: 02/14/2008] [Accepted: 02/19/2008] [Indexed: 10/22/2022]
Abstract
Aniline exposure causes toxicity to the spleen, which leads to a variety of sarcomas, and fibrosis appears to be an important preneoplastic lesion. However, early molecular mechanisms in aniline-induced toxicity to the spleen are not known. Previously, we have shown that aniline exposure results in iron overload and induction of oxidative stress in the spleen, which can cause transcriptional upregulation of fibrogenic/inflammatory cytokines via activation of oxidative stress (OS)-responsive signaling pathways. To test this mechanism, male SD rats were treated with aniline (1mmol/kg/day via gavage) for 7 days, an experimental condition that precedes the appearance of fibrosis. Significant increases in both NF-kappaB and AP-1 binding activity was observed in the nuclear extracts of splenocytes from aniline-treated rats as determined by ELISAs, and supported by Western blot data showing increases in p-IkappaBalpha, p-p65 and p-c-Jun. To understand the upstream signaling events which could account for the activation of NF-kappaB and AP-1, phosphorylation patterns of IkappaB kinases (IKKalpha and IKKbeta) and mitogen-activated protein kinases (MAPKs) were pursued. Our data showed remarkable increases in both p-IKKalpha and p-IKKbeta in the splenocytes from aniline-treated rats, suggesting their role in the phosphorylation of both IkappaBalpha and p65 subunits. Furthermore, aniline exposure led to activation of all three classes of MAPKs, as evident from increased phosphorylation of extracellular-signal-regulated kinase (ERK1/2), c-Jun N-terminal kinase (JNK1/2) and p38 MAPKs, which could potentially contribute to the observed activation of both AP-1 and NF-kappaB. Activation of upstream signaling molecules was also associated with simultaneous increases in gene transcription of cytokines IL-1, IL-6 and TNF-alpha. The observed sequence of events following aniline exposure could initiate a fibrogenic and/or tumorigenic response in the spleen.
Collapse
Affiliation(s)
- Jianling Wang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-0438, USA
| | | | | | | |
Collapse
|
43
|
Ogura H, Tsukumo Y, Sugimoto H, Igarashi M, Nagai K, Kataoka T. ERK and p38 MAP kinase are involved in downregulation of cell surface TNF receptor 1 induced by acetoxycycloheximide. Int Immunopharmacol 2008; 8:922-6. [DOI: 10.1016/j.intimp.2008.02.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2008] [Revised: 02/23/2008] [Accepted: 02/25/2008] [Indexed: 10/22/2022]
|
44
|
Trujillo-Murillo K, Rincón-Sánchez AR, Martínez-Rodríguez H, Bosques-Padilla F, Ramos-Jiménez J, Barrera-Saldaña HA, Rojkind M, Rivas-Estilla AM. Acetylsalicylic acid inhibits hepatitis C virus RNA and protein expression through cyclooxygenase 2 signaling pathways. Hepatology 2008; 47:1462-72. [PMID: 18393288 DOI: 10.1002/hep.22215] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
UNLABELLED It has been reported that salicylates (sodium salicylate and aspirin) inhibit the replication of flaviviruses, such as Japanese encephalitis virus and dengue virus. Therefore, we considered it important to test whether acetylsalicylic acid (ASA) had anti-hepatitis C virus (HCV) activity. To this end, we examined the effects of ASA on viral replication and protein expression, using an HCV subgenomic replicon cell culture system. We incubated Huh7 replicon cells with 2-8 mM ASA for different times and measured HCV-RNA and protein levels by northern blot, real-time polymerase chain reaction, and western analysis, respectively. We found that ASA had a suppressive effect on HCV-RNA and protein levels (nearly 58%). ASA-dependent inhibition of HCV expression was not mediated by the 5'-internal ribosome entry site or 3'-untranslated regions, as determined by transfection assays using bicistronic constructs containing these regulatory regions. However, we found that HCV-induced cyclooxygenase 2 (COX-2) messenger RNA and protein levels and activity and these effects were down-regulated by ASA, possibly by a nuclear factor kappa B-independent mechanism. We also observed that the ASA-dependent inhibition of viral replication was due in part to inhibition of COX-2 and activation of p38 and mitogen-activated protein kinase/extracellular signal-regulated kinase kinase 1/2 (MEK1/2) mitogen-activated protein kinases (MAPKs). Inhibition of these kinases by SB203580 and U0126, respectively, and by short interfering RNA silencing of p38 and MEK1 MAPK prevented the antiviral effect of ASA. Taken together, our findings suggest that the anti-HCV effect of ASA in the Huh7 replicon cells is due to its inhibitory effect on COX-2 expression, which is mediated in part by the activation of MEK1/2/p38 MAPK. CONCLUSION These findings suggest the possibility that ASA could be an excellent adjuvant in the treatment of chronic HCV infection.
Collapse
Affiliation(s)
- Karina Trujillo-Murillo
- Department of Biochemistry, School of Medicine and University Hospital, Autonomous University of Nuevo Leon, Monterrey, Nuevo Leon, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
45
|
von Brandenstein MG, Ngum Abety A, Depping R, Roth T, Koehler M, Dienes HP, Fries JWU. A p38-p65 transcription complex induced by endothelin-1 mediates signal transduction in cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1783:1613-22. [PMID: 18457675 DOI: 10.1016/j.bbamcr.2008.04.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 03/31/2008] [Accepted: 04/01/2008] [Indexed: 11/29/2022]
Abstract
Endothelin-1 is a powerful mitogen for various tumor and non-tumor cells. Its signaling cascade induces the inflammatory NF-kappaB complex, leading to expression of a number of target genes. In this context, MAPK p38 has been regarded as a potential phosphate donor for the p65 subunit of NF-kappaB. In the present study in HeLa cells, we have found that ET-1 induced signalling activates the NF-kappaB transcription complex (TC) in the nucleus at 6 h specifically via ET-A - but not ET-B receptor. The TC contains p65, p38 (alpha and beta) - binding to the NLS of p65 in the cytoplasm - as well as p50, but no IkappaBalpha. Specific p38 inhibition by SB203580 or by siRNA interferes markedly with gene expression of several target genes. Complex formation occurs in the cytoplasm, and both transcription factors transmigrate as a complex in the nucleus. Overexpression of p38, treatment with Chrysin, MG132, or dimethylformamide shows dependence of TC on p38 as partner. In other tumor cells lines studied, ET-1 activates TC, with p38 as an important complex partner of p65. TC-induction by ET-1 contains about twice the amount of p38 than by TNFalpha. Thus, p38 may be an additional therapeutic target to control inflammatory gene expression in tumor cells.
Collapse
|
46
|
Ogura H, Tsukumo Y, Sugimoto H, Igarashi M, Nagai K, Kataoka T. Ectodomain shedding of TNF receptor 1 induced by protein synthesis inhibitors regulates TNF-α-mediated activation of NF-κB and caspase-8. Exp Cell Res 2008; 314:1406-14. [DOI: 10.1016/j.yexcr.2008.01.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 01/22/2008] [Accepted: 01/23/2008] [Indexed: 10/22/2022]
|
47
|
Jimenez-Lopez JM, Wu D, Cederbaum AI. Synergistic toxicity induced by prolonged glutathione depletion and inhibition of nuclear factor-kappaB signaling in liver cells. Toxicol In Vitro 2008; 22:106-15. [PMID: 17920235 PMCID: PMC2234037 DOI: 10.1016/j.tiv.2007.08.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Revised: 08/17/2007] [Accepted: 08/20/2007] [Indexed: 01/01/2023]
Abstract
TNF-alpha, GSH depletion and CYP2E1 are factors that play an important role in alcoholic liver disease. Activation of NF-kappaB prevents hepatocyte damage caused by TNF-alpha. This work describes the effect of NF-kappaB inhibition on toxicities caused by GSH depletion or arachidonic acid (AA) treatment in liver cells, and evaluates the possible influence of CYP2E1 overexpression. Cells were exposed to the NF-kappaB inhibitor BAY11-7082, in the absence or presence of l-buthionine sulfoximine (BSO) to block GSH synthesis. BSO toxicity was higher in CYP2E1-expressing E47 HepG2 cells compared to control cells; the incubation with BAY11-7082 potentiated BSO toxicity in both cell lines comparably. Several other agents which suppress activation of NF-kappaB increased BSO toxicity in E47 cells. NF-kappaB inhibition, however, did not sensitize E47 cells to AA toxicity. Suppressing activity of NF-kappaB also potentiated BSO, but not AA toxicity, in isolated rat hepatocytes. BAY11-7082 plus BSO induced a greater p38 MAPK activation as compared to BAY11-7082 or BSO alone, and a p38 MAPK inhibitor protected against the synergistic toxicity. In summary, inhibition of NF-kappaB sensitizes liver cells to toxicity linked to GSH depletion, probably accelerating the processes of thiol homeostasis deregulation and induction of apoptosis through a mechanism mediated by p38 MAPK.
Collapse
Affiliation(s)
- Jose M Jimenez-Lopez
- Department of Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | |
Collapse
|
48
|
Abstract
BACKGROUND High-grade glioma remains one of the most difficult cancers to treat. Recent studies in oncology have identified a role of the ubiquitous enzyme, cyclooxygenase (Cox), especially cyclooxygenase-2 (COX-2) in cell proliferation, and its inhibition in cancer control, apoptosis, as well as synergy with other forms of therapy. The inhibitors of the Cox enzyme are well known as members of the nonsteroidal anti-inflammatory drug (NSAID) class of pharmaceuticals. METHODS In vitro and in vivo studies of different cancers expressing COX-2, including glioma studies, along with the few clinical trials that have been reported are reviewed to specifically identify the actions of these agents. RESULTS The anticancer effect of the COX-2 inhibitors may occur irrelevant of COX-2 expression, and it appears to be drug-specific, as well as dose-specific in different cancers. In combination with chemotherapeutic agents, the COX-2 inhibitors may have an additive, synergistic, or inhibitory effect on tumor growth. CONCLUSIONS As evaluations of this class of drugs begin in glioma, in vitro and in vivo data should be acquired to accurately predict which compounds will have an effect in controlling tumor growth and at which doses these should be used. The actual expression and inhibition of COX-2 may not always be relevant to the effects on tumor growth.
Collapse
Affiliation(s)
- Pamela New
- Department of Neurosurgery/Neuro-oncology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
49
|
Yang H, Chen J, Yang J, Qiao S, Zhao S, Yu L. Cyclophilin A is upregulated in small cell lung cancer and activates ERK1/2 signal. Biochem Biophys Res Commun 2007; 361:763-7. [PMID: 17678621 DOI: 10.1016/j.bbrc.2007.07.085] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2007] [Accepted: 07/17/2007] [Indexed: 12/17/2022]
Abstract
Cyclophilin A (CypA), a peptidyl-prolyl cis-trans isomerase (PPIase), was originally identified as the intracellular receptor for cyclosporin A (CsA). Recently, correlations of CypA with tumor pathogenesis have been studied. Here, we studied the expression of CypA and its receptor CD147 in several kinds of lung cancer cells as well as a normal lung cell and found that in H446 cell, a kind of small cell lung cancer cell, the expression are the highest. The exogeneous CypA protein can substantially stimulate H446 cell growth in dependence on its PPIase activity. We also showed that CypA protein can stimulate ERK1/2 signal in dose and time dependent manners and almost has no effect to p38 and JNK signals. Elucidation of the precise role of CypA in these pathways may lead to new targeted therapies for small cell lung cancer.
Collapse
Affiliation(s)
- Huirong Yang
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200433, PR China
| | | | | | | | | | | |
Collapse
|
50
|
Abe K, Miyoshi K, Muto T, Ruspita I, Horiguchi T, Nagata T, Noma T. Establishment and characterization of rat dental epithelial derived ameloblast-lineage clones. J Biosci Bioeng 2007; 103:479-85. [PMID: 17609165 DOI: 10.1263/jbb.103.479] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2007] [Accepted: 02/20/2007] [Indexed: 01/21/2023]
Abstract
Teeth are the hardest tissues covered with enamel produced by ameloblasts. The ameloblast differentiation is controlled by sequential epithelial-mesenchymal interactions during tooth morphogenesis. However, the molecular mechanism of ameloblast differentiation remains unclear. To address this question, we developed an in vitro assay system to evaluate the molecular mechanism of amelogenesis. First, we established dental epithelium-derived clones from 6-day-old rat incisors and established that cells of the clone SRE-G5 were the largest producers of amelogenin mRNA. Next, we analyzed the effects of several chemicals on the amelogenin expression in SRE-G5 cells. Only mitogen-activated protein kinase (MAPK) activators enhanced amelogenin mRNA expression. This finding corresponded to the immunohistochemical data showing the presence of phosphorylated forms of p38, c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinase (ERK) during ameloblast differentiation. To examine the roles of MAPK signals, we compared the effects of anisomycin and sodium salicylate on the expression of tooth-related differentiation markers. Both anisomycin and sodium salicylate induced amelogenin, Abcg2, and Bmp4 mRNA and down-regulated p75NGFR mRNA. On the other hand, ALP, ectodin, Bmp2 and Fgf8 mRNA were up-regulated only by anisomycin. These results indicate that MAPK signaling functions, at least in part, as the inducer of ameloblast differentiation.
Collapse
Affiliation(s)
- Kaori Abe
- Department of Molecular Biology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | | | | | | | | | | | | |
Collapse
|